EPM2A Antibody

Laforin, Mouse Anti Human
Shipped with Ice Packs
In Stock

Product Specs

Introduction
EPM2A, a dual-specificity phosphatase, interacts with polyribosomes and might play a role in regulating glycogen metabolism. Mutations in the EPM2A gene are associated with Lafora disease, a form of myoclonic epilepsy.
Formulation
The antibody is supplied at a concentration of 1 mg/ml in a solution of PBS at pH 7.4 with 0.1% sodium azide.
Storage Procedures
Store at 4°C for up to 1 month. For longer-term storage, keep at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody has a shelf life of 12 months when stored at -20°C and 1 month when stored at 4°C.
Applications
This EPM2A antibody has undergone testing using ELISA and Western blot analysis to ensure its specificity and reactivity. However, as applications can vary, it is recommended to titrate the antibody for optimal results in each specific experiment. For Western blot analysis, a dilution range of 1:1,000 to 1:2,000 is recommended, with a starting dilution of 1:1,000.
Synonyms
Laforin, Lafora PTPase, LAFPTPase, EPM2A, EPM2, MELF, epilepsy progressive myoclonus type 2A Lafora disease.
Purification Method
EPM2A antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
P3F3AT.
Immunogen
Anti-human EPM2A mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human EPM2A amino acids 243-331 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and κ light chain.

Q&A

What is EPM2A and why is it important in scientific research?

EPM2A encodes a dual specificity phosphatase that has emerged as a significant protein in both neurological disorders and cancer research. This protein, also known as laforin, was originally identified in Lafora disease, a progressive form of myoclonic epilepsy . Recent studies indicate its importance extends to various cancers where it serves as a potential biomarker and prognostic indicator . The significance of EPM2A in scientific research stems from its involvement in multiple cellular processes including protein targeting to endoplasmic reticulum, cotranslational protein targeting to membrane, and participation in oxidoreductase activity and oxidative phosphorylation . Understanding EPM2A's functions provides insights into both disease mechanisms and potential therapeutic approaches.

What types of EPM2A antibodies are available for research applications?

Several types of EPM2A antibodies are available to researchers, varying in host species, clonality, and target epitopes. Polyclonal antibodies raised in rabbits that target various regions of human EPM2A are commonly used for multiple applications . Monoclonal antibodies, such as clone 6C6 from mouse hosts, provide high specificity for particular epitopes (e.g., AA 101-199) . Researchers can select from antibodies targeting different regions including the C-terminal domain, internal regions, or specific amino acid sequences (e.g., AA 1-331, AA 244-331, AA 80-130) . These antibodies come in different forms including unconjugated versions for flexibility in detection methods and conjugated versions like biotinylated antibodies for specialized applications . Selection should be based on the specific experimental requirements, target species, and application methodology.

What are the common applications for EPM2A antibodies in research?

EPM2A antibodies can be utilized across multiple research applications, with Western Blotting (WB), Enzyme-Linked Immunosorbent Assay (ELISA), and Immunohistochemistry (IHC) being the most common . For tissue localization studies, researchers frequently employ IHC to examine EPM2A expression patterns in normal versus pathological samples, as demonstrated in prostate cancer tissue microarrays . Immunofluorescence (IF) techniques allow for visualization of subcellular localization and co-localization studies with other proteins of interest . For protein-protein interaction studies, Immunoprecipitation (IP) with EPM2A antibodies can be valuable . When selecting an EPM2A antibody, researchers should confirm the validated applications listed by manufacturers, as not all antibodies perform equally across all methodologies . For quantitative expression analysis, ELISA provides a reliable method for measuring EPM2A levels in biological samples .

How should I validate an EPM2A antibody before using it in my experiments?

Proper validation of EPM2A antibodies is crucial for experimental reliability. Begin with positive and negative controls—use samples with known EPM2A expression (such as specific cell lines) and compare against knockout/knockdown models or tissues known to lack EPM2A expression . Western blotting should be performed to confirm the antibody detects a band of the correct molecular weight (~37 kDa for the main isoform of human EPM2A) . For IHC applications, compare staining patterns with published literature, such as the differential expression patterns observed between normal and tumor tissues in prostate cancer studies . Antibody specificity can be further confirmed through peptide competition assays, where pre-incubation with the immunizing peptide should abolish specific signals. Cross-reactivity testing is important if working with multiple species; while many EPM2A antibodies recognize human, rat, and mouse proteins, species-specific differences in reactivity exist . Finally, consider validating across multiple techniques (WB, IHC, IF) to ensure consistent performance across your intended applications.

How can I optimize immunohistochemistry protocols for EPM2A detection in different tissue types?

Optimizing IHC protocols for EPM2A detection requires careful consideration of tissue-specific characteristics. For formalin-fixed paraffin-embedded (FFPE) tissues, heat-induced epitope retrieval is typically necessary, with citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) commonly used . Based on prostate cancer studies, a semi-quantitative scoring system combining positively stained region scores (0 for negative, 1 for 1%-10%, 2 for 11%-50%, 3 for 51%-80%, and 4 for >80%) with immunostaining intensity scores (0 for no staining, 1 for weak, 2 for mild, and 3 for strong) provides reliable assessment of EPM2A expression . For multi-color IHC when investigating EPM2A alongside immune markers like PD-1, sequential antibody incubation with appropriate blocking steps is crucial to prevent cross-reactivity . Tissue-specific optimization is important—prostate cancer tissues may require different conditions than neurological tissues where EPM2A was originally studied. Primary antibody dilution should be empirically determined, typically starting at 1:100-1:500 for commercial antibodies . Inclusion of positive control tissues with known EPM2A expression and negative controls (primary antibody omission) in each experiment ensures protocol reliability. For quantitative analysis, digital image analysis tools can provide objective measurement of EPM2A expression intensity and distribution patterns.

What are the known challenges in detecting different EPM2A isoforms, and how can they be addressed?

Detection of specific EPM2A isoforms presents significant challenges due to sequence homology and structural similarities. Human EPM2A gene contains multiple exons that undergo alternative splicing, resulting in at least 9 known isoforms with different subcellular localizations and functions . To distinguish between isoforms, researchers should select antibodies raised against isoform-specific regions—for instance, antibodies targeting AA 1-331 (Isoform 9) versus those targeting other amino acid sequences . Western blotting optimization is crucial: using gradient gels (4-20%) can help separate isoforms with similar molecular weights, while longer running times and lower voltage improve band separation. For mass spectrometry-based isoform identification, tryptic digestion followed by analysis of isoform-specific peptides provides definitive discrimination. RNA-level validation through RT-PCR with isoform-specific primers should complement protein-level detection. When studying localization, subcellular fractionation before Western blotting helps confirm isoform-specific distribution patterns, as some EPM2A isoforms localize to the nucleus while others remain cytoplasmic. Co-immunoprecipitation experiments with isoform-specific antibodies can reveal differential protein interaction networks. Researchers should also be aware that isoform expression varies between tissue types and disease states, requiring careful selection of appropriate positive controls for each isoform being studied.

How can EPM2A antibodies be used to investigate the relationship between EPM2A expression and immune infiltration in cancer?

EPM2A antibodies provide valuable tools for investigating the relationship between EPM2A expression and immune cell infiltration in cancer tissues. Multiplex immunofluorescence or immunohistochemistry techniques can be employed to simultaneously detect EPM2A alongside immune cell markers for B cells, T cells, macrophages, and other immune cell populations . Research in prostate cancer has shown that high EPM2A expression correlates with increased infiltration of activated B cells, immature B cells, macrophages, mast cells, effector memory CD8 T cells, T follicular cells, natural killer cells, and various T helper cell subsets . To quantify this relationship, tissue sections should be analyzed using digital pathology platforms capable of cell-specific quantification within the tumor microenvironment. Single-cell suspensions from fresh tumor samples can be analyzed by flow cytometry after staining with EPM2A antibodies and immune cell markers to quantify associations at the single-cell level. Co-localization studies combining EPM2A antibodies with antibodies against immune checkpoint molecules (PD-1, PD-L1, PD-L2) are particularly valuable given the positive correlation between EPM2A expression and these checkpoint molecules in prostate cancer . For mechanistic insights, in vitro co-culture systems using EPM2A-expressing cancer cells with various immune cell populations can help determine if EPM2A directly influences immune cell recruitment or activation. Correlation analyses between EPM2A expression and established immune gene signatures can provide additional evidence for EPM2A's role in modulating the tumor immune microenvironment.

How can EPM2A antibodies be used to investigate the relationship between EPM2A expression and therapy response in cancer patients?

EPM2A antibodies play a crucial role in investigating connections between EPM2A expression and therapy response in cancer. For retrospective studies, immunohistochemistry on pre-treatment biopsy specimens using validated EPM2A antibodies can establish baseline expression levels before correlating with treatment outcomes . Research in prostate cancer demonstrated that patients with low EPM2A expression exhibited increased sensitivity to cisplatin, paclitaxel, and bicalutamide, as evidenced by lower IC50 values . For prospective investigations, sequential biopsies analyzed with EPM2A antibodies can track expression changes during treatment, providing insights into adaptive responses. In vitro drug sensitivity assays using cancer cell lines with varying EPM2A expression levels (achieved through overexpression or knockdown) allow for controlled evaluation of EPM2A's impact on drug response mechanisms. For immunotherapy response prediction, multiplex immunohistochemistry combining EPM2A antibodies with immune checkpoint markers (PD-1, PD-L1, PD-L2) is particularly valuable given the positive correlation observed between EPM2A expression and these markers in prostate cancer . Patient-derived xenograft models with differential EPM2A expression can be treated with various therapeutic agents to validate in vitro findings in a more physiologically relevant system. For clinical application, developing immunohistochemistry-based diagnostic assays with carefully optimized cutoff values for EPM2A expression could help stratify patients into therapy-responsive groups. Integration of EPM2A expression data with other biomarkers into multiparametric prediction models may enhance accuracy of therapy response predictions beyond what can be achieved with EPM2A status alone.

How can I perform co-immunoprecipitation experiments using EPM2A antibodies to identify novel protein interactions?

Co-immunoprecipitation (Co-IP) using EPM2A antibodies requires careful optimization to identify genuine interaction partners. Begin by selecting an EPM2A antibody validated for immunoprecipitation applications, such as rabbit polyclonal antibodies targeting amino acids 1-331 . Cell lysate preparation is critical—use mild lysis buffers (containing 1% NP-40 or 0.5% Triton X-100) to preserve protein-protein interactions, with protease and phosphatase inhibitors to prevent degradation during processing. Pre-clearing the lysate with protein A/G beads reduces non-specific binding. When coupling the EPM2A antibody to beads, determine optimal antibody-to-bead ratios empirically (typically 2-5 μg antibody per 50 μl bead slurry). For negative controls, include isotype-matched control antibodies and, ideally, lysates from EPM2A-knockout or knockdown cells. Following immunoprecipitation, thorough washing (4-5 washes) with decreasing salt concentrations helps eliminate non-specific interactions while preserving genuine ones. For protein identification, either targeted Western blotting with antibodies against suspected interaction partners or unbiased mass spectrometry analysis can be employed. When using mass spectrometry, compare proteins identified in EPM2A immunoprecipitates against control immunoprecipitates and establish stringent enrichment thresholds. Confirmation of novel interactions should be performed through reciprocal Co-IP (using antibodies against the identified partner to pull down EPM2A) and orthogonal methods such as proximity ligation assays or FRET analyses. Based on pathway enrichment analyses from prostate cancer studies, investigating interactions with proteins involved in endoplasmic reticulum targeting, ribosome structural constituents, and oxidative phosphorylation may be particularly informative .

How can EPM2A antibodies be incorporated into high-throughput screening approaches for biomarker discovery?

Incorporating EPM2A antibodies into high-throughput screening facilitates efficient biomarker discovery across large sample sets. Tissue microarrays (TMAs) containing hundreds of tumor samples with matched normal tissues can be simultaneously screened using validated EPM2A antibodies, similar to the approach used in prostate cancer studies . Automated immunohistochemistry platforms ensure consistent staining conditions across all samples, while digital pathology systems with machine learning algorithms can quantify EPM2A expression, reducing subjective interpretation. For protein array approaches, EPM2A antibodies can be applied to reverse-phase protein arrays (RPPAs) containing lysates from multiple patient samples to assess expression correlation with other potential biomarkers. Multiplexed approaches are particularly valuable—Tyramide Signal Amplification (TSA) allows simultaneous detection of EPM2A alongside multiple other markers (up to 7-10) on the same tissue section, enabling comprehensive profiling of the tumor microenvironment. Integration with genomic and transcriptomic data is essential; correlating EPM2A protein expression with RNA-seq data can identify transcriptional mechanisms regulating EPM2A levels and associated molecular pathways . For functional screening, CRISPR-Cas9 libraries targeting genes in EPM2A-related pathways, followed by EPM2A antibody-based readouts, can identify modulators of EPM2A expression or function. High-content imaging systems combining EPM2A antibody staining with cellular phenotype assessment provide insights into functional consequences of EPM2A expression variation. Finally, analysis pipelines should implement machine learning algorithms to identify complex relationships between EPM2A expression patterns and clinical outcomes, potentially uncovering novel biomarker signatures with greater predictive power than EPM2A alone.

What are the best practices for using EPM2A antibodies in flow cytometry and cell sorting applications?

Optimizing EPM2A antibodies for flow cytometry and cell sorting requires special considerations due to EPM2A's predominantly intracellular localization. Cell fixation and permeabilization are essential steps—paraformaldehyde fixation (2-4%) followed by permeabilization with saponin (0.1-0.5%) or Triton X-100 (0.1-0.3%) typically provides good results for intracellular proteins like EPM2A. When selecting antibodies, prioritize those specifically validated for flow cytometry or those with demonstrated specificity in immunofluorescence applications . Fluorophore selection should consider the instrument configuration and other markers in your panel; bright fluorophores (PE, APC) are advantageous for detecting proteins with moderate expression levels like EPM2A. Titration experiments are crucial to determine optimal antibody concentration, typically starting with manufacturer's recommendations and testing 2-fold dilutions above and below. Controls must include unstained cells, isotype controls matched to the EPM2A antibody's host species and isotype, and ideally, EPM2A-knockout or knockdown cells as negative controls. For multiparameter analysis investigating EPM2A alongside immune markers (reflecting its association with immune infiltration in cancer ), careful panel design with appropriate compensation controls is essential. When using EPM2A expression for cell sorting, setting gates requires proper controls, and sorted populations should be reanalyzed to confirm enrichment. For downstream applications following sorting, verify that fixed/permeabilized cells remain suitable for your intended analyses. Finally, consider whether examining EPM2A in conjunction with cell cycle markers might provide insights into its varied expression during different cell cycle phases, potentially explaining some of its tumor-suppressive functions observed in prostate cancer .

How should researchers interpret apparent contradictions in EPM2A expression patterns across different cancer types?

Interpreting contradictory EPM2A expression patterns across cancer types requires careful consideration of biological context and methodological differences. First, determine whether contradictions reflect genuine biological differences or methodological variations—standardize analysis methods across datasets, including antibody selection, staining protocols, and scoring systems . Cancer type-specific roles for EPM2A likely exist; in prostate cancer, EPM2A functions as a tumor suppressor with decreased expression in tumors , but this pattern may differ in other cancers due to tissue-specific signaling networks. Consider cancer subtype heterogeneity—expression patterns that appear contradictory might reflect different molecular subtypes within broader cancer categories. Importantly, investigate EPM2A at multiple biological levels—while protein expression measured by antibodies might decrease, genetic alterations or mRNA expression might show different patterns. Pathway context is crucial; examine EPM2A in relation to associated pathways identified through enrichment analysis, such as protein targeting to endoplasmic reticulum, oxidative phosphorylation, and glycolysis . The tumor microenvironment likely influences EPM2A expression and function—prostate cancers with high EPM2A expression showed increased immune cell infiltration and checkpoint molecule expression , suggesting immunological context may explain some apparent contradictions. Technical factors including antibody epitope accessibility, isoform specificity, and post-translational modifications can create artifactual differences . Finally, consider functional consequences rather than just expression levels—the same protein may have context-dependent functions across different tissues, potentially explaining why altered expression has different implications in various cancer types.

How can researchers integrate EPM2A protein expression data with transcriptomic and genomic datasets for comprehensive analysis?

Integrating EPM2A protein expression data with transcriptomic and genomic datasets enables comprehensive multi-omic analysis that provides deeper biological insights. Start by normalizing data across platforms—for protein expression quantified by immunohistochemistry, convert semi-quantitative scores to continuous values that can be compared with gene expression data . Correlation analysis between EPM2A protein and mRNA levels helps identify post-transcriptional regulation mechanisms; discrepancies suggest regulatory processes like miRNA targeting or protein degradation pathways. Integration with mutation data can reveal whether specific genomic alterations affect EPM2A expression; this is particularly relevant given EPM2A's dual role in neurological disorders and cancer . For pathway-level integration, perform gene set enrichment analysis (GSEA) using transcriptomic data from samples stratified by EPM2A protein expression, as done in prostate cancer studies that identified altered pathways in high versus low EPM2A expression groups . Network analysis incorporating protein-protein interaction databases with EPM2A as a central node can reveal functional relationships between genomic alterations and protein expression changes. To integrate with epigenomic data, correlate EPM2A protein expression with methylation status of its promoter region to assess epigenetic regulation. Causal inference methods like Mendelian randomization can help determine whether genetic variants influencing EPM2A expression have downstream effects on phenotypes or disease outcomes. For visualization, multi-omic data browsers displaying EPM2A protein expression alongside genomic alterations, transcriptomic changes, and clinical data provide intuitive exploration of complex relationships. Finally, machine learning approaches—particularly ensemble methods combining features from multiple omic layers—can identify complex patterns associated with EPM2A expression that might not be apparent in single-platform analyses.

What are common troubleshooting strategies for weak or inconsistent EPM2A antibody signals in Western blotting?

When encountering weak or inconsistent EPM2A antibody signals in Western blotting, systematic troubleshooting is essential. Start by optimizing protein extraction—EPM2A may distribute between different cellular compartments, so using appropriate lysis buffers (RIPA or NP-40 based) with complete protease inhibitor cocktails prevents degradation . For loading controls, BCA or Bradford assays ensure equal protein concentration across samples. Sample preparation significantly impacts detection—fresh samples typically yield better results than frozen-thawed materials, and reducing agent concentration in sample buffer may need adjustment if EPM2A contains disulfide bonds affecting epitope accessibility. Transfer efficiency should be verified using reversible total protein stains like Ponceau S before immunoblotting. Primary antibody optimization is crucial—titrate concentrations (typically testing 1:500-1:5000 dilutions) and extend incubation time (overnight at 4°C often improves signal-to-noise ratio) . If signals remain weak, signal enhancement systems like biotin-streptavidin amplification or highly sensitive chemiluminescent substrates can improve detection. For inconsistent signals across experiments, standardize all protocols and reagents, and consider preparing larger antibody aliquots to minimize freeze-thaw cycles. Epitope masking may occur if EPM2A undergoes post-translational modifications or interactions with other proteins—stronger denaturing conditions or phosphatase treatment before electrophoresis might help. Batch effects between different lots of the same antibody can be significant; maintain records of lot numbers and perform side-by-side comparisons when changing lots . Finally, if particular EPM2A antibodies consistently underperform, test alternatives targeting different epitopes, as some regions may be more accessible or immunogenic than others in Western blotting applications .

How can researchers address non-specific binding issues when using EPM2A antibodies in complex tissue samples?

Addressing non-specific binding of EPM2A antibodies in complex tissues requires a multi-faceted approach. Initially, optimize blocking conditions—test various blocking agents (BSA, milk, serum, commercial blockers) at different concentrations (3-5%) and times (1-2 hours) to identify combinations that minimize background while preserving specific signals . For IHC applications, tissue-specific autofluorescence or endogenous peroxidase activity can contribute to background—implement appropriate quenching steps (hydrogen peroxide treatment for peroxidase activity, sodium borohydride for aldehyde-induced autofluorescence) before antibody incubation . Antibody dilution optimization is critical; perform systematic titration experiments starting from manufacturer's recommendations and create signal-to-noise curves to determine optimal concentration . Extensive washing between steps with appropriate detergents (0.05-0.1% Tween-20) helps remove weakly bound antibodies. Consider antibody format—F(ab) or F(ab')2 fragments may provide lower background than complete IgG in tissues rich in Fc receptors. Pre-adsorption of primary antibodies with tissue homogenates from irrelevant species can reduce cross-reactivity. For multiple antigen detection, careful selection of primary antibodies from different host species minimizes cross-reactivity between detection systems. When these approaches prove insufficient, try alternative EPM2A antibodies targeting different epitopes—polyclonal antibodies targeting non-overlapping regions can be useful for confirmation by showing concordant staining patterns . Always include appropriate controls: isotype controls at matching concentrations, tissue sections lacking primary antibody treatment, and ideally, tissues from EPM2A knockout models or those treated with EPM2A-targeting siRNA. For quantitative analyses, background subtraction algorithms in image analysis software can help mitigate effects of residual non-specific binding.

What strategies can help distinguish between closely related proteins when using EPM2A antibodies?

Distinguishing EPM2A from closely related proteins requires strategic approaches to ensure antibody specificity. Start with bioinformatic analysis of protein sequence homology to identify regions unique to EPM2A—this knowledge guides selection of antibodies targeting EPM2A-specific epitopes rather than conserved domains shared with related phosphatases . Comprehensive validation using positive and negative controls is essential: cell lines or tissues with confirmed EPM2A expression serve as positive controls, while EPM2A knockout/knockdown models provide definitive negative controls that should show absence of signal . Western blotting with careful attention to molecular weight is valuable—EPM2A has a characteristic molecular weight (~37 kDa for the main isoform), distinguishable from related proteins . Peptide competition assays, where primary antibody is pre-incubated with excess immunizing peptide before application to samples, should abolish specific EPM2A signals while leaving non-specific binding to related proteins unaffected. For challenging cases, two-dimensional gel electrophoresis before Western blotting separates proteins by both molecular weight and isoelectric point, providing enhanced discrimination. Immunoprecipitation followed by mass spectrometry analysis offers definitive identification, confirming that the antibody-captured protein is indeed EPM2A rather than a related protein. Sequential immunodepletion, where samples are first cleared of EPM2A using a validated antibody before probing with the antibody under investigation, helps determine whether signals derive from EPM2A or cross-reactive proteins. Finally, parallel testing with multiple antibodies targeting different EPM2A epitopes provides confidence in specificity—genuine EPM2A detection should show concordant results across antibodies, while cross-reactivity patterns typically differ between antibodies .

How might EPM2A antibodies contribute to understanding the dual role of EPM2A in neurological disorders and cancer?

EPM2A antibodies will be instrumental in elucidating the seemingly disparate roles of EPM2A in neurological disorders and cancer. Comparative immunohistochemistry studies using validated EPM2A antibodies across brain tissues affected by Lafora disease and various cancer types can reveal tissue-specific expression patterns and subcellular localization differences that might explain contextual functions . Phospho-specific EPM2A antibodies that distinguish between different phosphorylation states could identify regulatory mechanisms that switch EPM2A between its neurological and oncological roles. Co-immunoprecipitation experiments paired with mass spectrometry can map tissue-specific protein interaction networks, potentially revealing how the same protein serves different functions through distinct binding partners in neural versus cancer cells . For mechanistic studies, proximity labeling approaches (BioID, APEX) combined with EPM2A antibodies for verification can identify proteins in close physical proximity to EPM2A in different cellular contexts. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using EPM2A antibodies might uncover unexpected nuclear functions if EPM2A influences gene expression differently in neuronal versus cancer cells. In animal models, tissue-specific conditional knockouts followed by rescue experiments with wild-type or mutant EPM2A can determine which functional domains are critical in each disease context, with EPM2A antibodies confirming expression levels and localization patterns. Single-cell analyses using EPM2A antibodies can reveal cell-type specific expression and potential roles within complex tissues. Finally, therapeutic development targeting EPM2A must consider its dual nature—EPM2A antibodies will be essential in screening assays to identify compounds that modulate EPM2A function in a tissue-specific manner, potentially normalizing function in neurological disorders while enhancing tumor-suppressive activity in cancers .

What role might EPM2A antibodies play in developing targeted therapies or companion diagnostics for cancer treatment?

EPM2A antibodies could significantly advance both targeted therapy development and companion diagnostics for personalized cancer treatment strategies. Standardized immunohistochemistry assays using validated EPM2A antibodies could serve as companion diagnostics to stratify patients for treatment selection, building on research showing that low EPM2A expression correlates with increased sensitivity to cisplatin, paclitaxel, and bicalutamide in prostate cancer . For immunotherapy selection, EPM2A assessment alongside PD-1, PD-L1, and PD-L2 expression might identify patients likely to benefit from immune checkpoint inhibitors, given the positive correlation between EPM2A and these markers . Beyond diagnostics, EPM2A antibody-drug conjugates (ADCs) could deliver cytotoxic agents specifically to cancer cells expressing EPM2A, though this approach requires careful consideration of expression in normal tissues. Bispecific antibodies linking EPM2A-expressing cancer cells with immune effectors represent another therapeutic possibility, particularly relevant given EPM2A's association with immunocyte infiltration . For monitoring treatment response, serial liquid biopsies analyzed with highly sensitive EPM2A antibody-based assays could track circulating tumor cells or exosomes expressing EPM2A. In drug discovery, phenotypic screening assays incorporating EPM2A antibodies can identify compounds that modulate EPM2A expression or function, particularly targeting the phosphatase activity central to its tumor-suppressive role . High-throughput screening of small molecule libraries for compounds that increase EPM2A expression or activity might yield novel therapeutic candidates, with EPM2A antibodies providing the detection method. Finally, structure-function studies combining X-ray crystallography or cryo-EM of EPM2A with epitope mapping using diverse antibodies could reveal druggable pockets for rational design of EPM2A modulators, potentially establishing entirely new treatment avenues for cancers where EPM2A plays a significant role .

How can emerging antibody technologies enhance our understanding of EPM2A biology and function?

Emerging antibody technologies offer unprecedented opportunities to advance EPM2A research beyond traditional applications. Single-domain antibodies (nanobodies) derived from camelid species provide superior tissue penetration and access to EPM2A epitopes that might be inaccessible to conventional antibodies, enabling more comprehensive mapping of functional domains . Intrabodies—antibodies designed for intracellular expression—could allow real-time visualization of endogenous EPM2A in living cells, providing insights into dynamic processes including trafficking between subcellular compartments relevant to its diverse functions . For spatial biology, in situ proximity ligation assays using EPM2A antibodies can visualize protein-protein interactions within their native cellular context, helping map the EPM2A interactome in different tissues and disease states. Antibody-based biosensors incorporating Förster resonance energy transfer (FRET) or bioluminescence resonance energy transfer (BRET) technology could monitor EPM2A conformational changes or interactions in real-time, potentially revealing regulatory mechanisms. Mass cytometry (CyTOF) using metal-conjugated EPM2A antibodies enables high-dimensional analysis of EPM2A expression alongside dozens of other markers at single-cell resolution, facilitating comprehensive profiling of cellular heterogeneity in complex tissues . For proteomics, antibody arrays containing multiple EPM2A antibodies targeting different epitopes or modifications enable multiplexed analysis of EPM2A variants across numerous samples simultaneously. DNA-barcoded antibody technology combines the specificity of EPM2A antibodies with the quantitative power of next-generation sequencing, allowing digital counting of EPM2A proteins with exceptional sensitivity. Finally, computational antibody design using machine learning approaches could generate novel EPM2A antibodies with enhanced specificity for particular isoforms or post-translational modifications, addressing current limitations in distinguishing these variants . Together, these emerging technologies promise to transform our understanding of EPM2A biology and its roles in both neurological disorders and cancer.

Product Science Overview

Introduction

Laforin is a protein encoded by the EPM2A gene and is primarily known for its role in Lafora disease, a fatal autosomal recessive neurodegenerative disorder. Lafora disease is characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies. These inclusions are found in the cytoplasm of cells in the central nervous system and other organs . Laforin is a member of the dual-specificity protein phosphatase family and plays a crucial role in regulating glycogen metabolism and autophagy .

Preparation Methods

The preparation of mouse anti-human laforin antibodies involves several steps:

  1. Antigen Preparation: The human laforin protein is expressed and purified using recombinant DNA technology. The purified protein serves as the antigen for immunization.
  2. Immunization: Mice are immunized with the purified human laforin protein. The immunization process typically involves multiple injections over several weeks to elicit a strong immune response.
  3. Hybridoma Production: Spleen cells from the immunized mice are fused with myeloma cells to create hybridoma cells. These hybridoma cells are screened for the production of antibodies specific to human laforin.
  4. Antibody Purification: The antibodies produced by the hybridoma cells are purified using techniques such as protein A/G affinity chromatography.
Chemical Reactions Analysis

Laforin’s primary function is to dephosphorylate glycogen, preventing the formation of poorly branched glycogen that accumulates as Lafora bodies . The protein’s phosphatase activity is crucial for maintaining normal glycogen metabolism. However, recent studies have shown that the phosphatase activity of laforin is not essential for rescuing mice from Lafora disease . Instead, laforin’s role in regulating autophagy and its interaction with the E3 ubiquitin ligase malin are critical for preventing the accumulation of Lafora bodies .

Laforin regulates autophagy via the mammalian target of rapamycin (mTOR) kinase-dependent pathway . The loss of laforin impairs autophagy, leading to the accumulation of autophagy substrates and contributing to cell stress and death . The interaction between laforin and malin is essential for controlling abnormal glycogen accumulation through intracellular proteolytic systems .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.